1
|
Illingworth EJ, Rychlik KA, Maertens A, Sillé FCM. Sex-specific transcriptomic effects of low-dose inorganic arsenic exposure on bone marrow-derived macrophages. Toxicology 2025; 510:153988. [PMID: 39515575 DOI: 10.1016/j.tox.2024.153988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/19/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Both tissue-resident macrophages and monocytes recruited from the bone marrow that transform into tissue-resident cells play critical roles in mediating homeostasis as well as in the pathology of inflammatory diseases. Inorganic arsenic (iAs) is the most common drinking water contaminant worldwide and represents a major public health concern. There are numerous diseases caused by iAs exposure in which macrophages are involved, including cardiovascular disease, cancer, and increased risk of (respiratory) infectious diseases. Notably, prenatal iAs exposure is also associated with negative birth outcomes and developmental immunotoxicity (DIT) contributing to long-term adverse outcomes of these immune-related diseases. Therefore, understanding the effects of iAs exposure on macrophages, particularly during immune development or tissue injury and inflammation, can help us better grasp the full range of arsenic immunotoxicity and better design therapeutic targets for iAs-induced diseases particularly in exposed populations. In contrast to prior published studies which often only focused on the effect of iAs on mature macrophages after development, in this study, we analyzed the transcriptome of M0-, M1- and M2-polarized male and female murine bone marrow-derived macrophages (BMDMs) which were exposed to iAs during the differentiation phase, as a model to study iAs (developmental) immunotoxicity. We identified differentially expressed genes by iAs in a sex- and stimulation-dependent manner and used bioinformatics tools to predict protein-protein interactions, transcriptional regulatory networks, and associated biological processes. Overall, our data suggest that M1-stimulated, especially female-derived, BMDMs are most susceptible to iAs exposure during differentiation. Most notably, we observed significant downregulation of major proinflammatory transcription factors, like IRF8, and its downstream targets, as well as genes encoding proteins involved in pattern recognition and antigen presentation, such as TLR7, TLR8, and H2-D1, potentially providing causal insight regarding the role of (early-life) arsenic exposure in perturbing immune responses to infectious diseases. We also observed significant downregulation of genes involved in processes crucial to coordinating a proinflammatory response including leukocyte migration, differentiation, and cytokine and chemokine production and response. Finally, we discovered that 24 X-linked genes were dysregulated in iAs-exposed female stimulation groups compared to only 3 across the iAs-exposed male stimulation groups. These findings elucidate the potential mechanisms underlying the sex-differential iAs-associated immune-related disease risk.
Collapse
Affiliation(s)
- Emily J Illingworth
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Kristal A Rychlik
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA; Public Health Program, School of Health Professions, Mayborn College of Health Sciences, University of Mary Hardin-Baylor, Belton, TX, USA
| | - Alexandra Maertens
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Fenna C M Sillé
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
2
|
Lorenzo-Anota HY, Gómez-Cantú JM, Vázquez-Garza E, Bernal-Ramirez J, Chapoy-Villanueva H, Mayolo-Deloisa K, Benavides J, Rito-Palomares M, Lozano O. Disulfiram-Loaded Nanoparticles Inhibit Long-Term Proliferation on Preadipocytes. Int J Nanomedicine 2024; 19:13301-13318. [PMID: 39679252 PMCID: PMC11645963 DOI: 10.2147/ijn.s467909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 11/03/2024] [Indexed: 12/17/2024] Open
Abstract
Introduction Disulfiram (DSF) reduces insulin resistance and weight gain in obese mice. However, the effect on adipose tissue is unexplored due to their high instability under physiological conditions, limiting clinical applications. Thus, it is meaningful to develop a DSF carrier for sustained release to adipose tissue. We optimized the synthesis of poly-ε-caprolactone (PCL) nanoparticles (NPs) loaded with DSF and analyzed their effect on adipose tissue cells in vitro. Methods The NPs were synthesized by nanoprecipitation method, varying its solvent, either acetone or acetone/dichloromethane (60:40) (v/v), and ratio PCL:DSF (w/w) 1:2, 1:1, 2:1 and, 1:0; finding the best condition was obtained with acetone/dichloromethane solvent mixture and 2:1 PCL:DSF. Then, NPs toxicity was analyzed on adipose cells (preadipocytes, white-like adipocytes, and macrophages) assessing association and internalization, cell viability, and cell death mechanism. Results NPs were spherical with a particle size distribution of 203.2 ± 29.33 nm, a ζ-potential of -20.7 ± 4.58 mV, a PDI of 0.296 ± 0.084, and a physical drug loading of 18.6 ± 5.80%. Sustained release was observed from 0.5 h (10.94 ± 2.38%) up to 96 h (91.20 ± 6.03%) under physiological conditions. NPs internalize into macrophages, white-like adipocytes and preadipocytes without modifying cell viability on white-like adipocytes and macrophages. Preadipocytes reduce cell viability, inducing mitochondrial damage, increased mitochondrial reactive oxygen species production and loss of mitochondrial membrane potential, leading to effector caspases 3/7 cleaved, resulting in apoptosis. Finally, long-term proliferation inhibition was observed, highlighting the bioequivalent effect of PCL-DSF NPs compared to free DSF. Conclusion Our data demonstrated the biological interaction of PCL NPs with adipose cells in vitro. The selective cytotoxicity of DSF towards preadipocytes resulted in milder effects when it was delivered nanoencapsulated compared to the free drug. These results suggest promising pharmacological alternatives for DSF long-term delivery on adipose tissue.
Collapse
Affiliation(s)
- Helen Yarimet Lorenzo-Anota
- Tecnologico de Monterrey, Institute for Obesity Research, Monterrey, México
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, México
| | | | | | | | - Héctor Chapoy-Villanueva
- Tecnologico de Monterrey, Institute for Obesity Research, Monterrey, México
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, México
| | - Karla Mayolo-Deloisa
- Tecnologico de Monterrey, Institute for Obesity Research, Monterrey, México
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Monterrey, México
| | - Jorge Benavides
- Tecnologico de Monterrey, Institute for Obesity Research, Monterrey, México
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Monterrey, México
| | - Marco Rito-Palomares
- Tecnologico de Monterrey, Institute for Obesity Research, Monterrey, México
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, México
| | - Omar Lozano
- Tecnologico de Monterrey, Institute for Obesity Research, Monterrey, México
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, México
| |
Collapse
|
3
|
Carrazana R, Espinoza F, Ávila A. Mechanistic perspective on the actions of vitamin a in autism spectrum disorder etiology. Neuroscience 2024; 554:72-82. [PMID: 39002756 DOI: 10.1016/j.neuroscience.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 05/07/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
Vitamin A (VA) has many functions in the body, some of which are key for the development and functioning of the nervous system, while some others might indirectly influence neural function. Both hypovitaminosis and hypervitaminosis A can lead to clinical manifestations of concern for individuals and for general global health. Scientific evidence on the link between VA and autism spectrum disorder (ASD) is growing, with some clinical studies and accumulating results obtained from basic research using cellular and animal models. Remarkably, it has been shown that VA deficiency can exacerbate autistic symptomatology. In turn, VA supplementation has been shown to be able to improve autistic symptomatology in selected groups of individuals with ASD. However, it is important to recognize that ASD is a highly heterogeneous condition. Therefore, it is important to clarify how and when VA supplementation can be of benefit for affected individuals. Here we delve into the relationship between VA and ASD, discussing clinical observations and mechanistic insights obtained from research on selected autistic syndromes and laboratory models to advance in defining how the VA signaling pathway can be exploited for treatment of ASD.
Collapse
Affiliation(s)
- Ramón Carrazana
- Neurodevelopmental Biology Unit, Biomedical Sciences Research Laboratory, Basic Sciences Department, Faculty of Medicine, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - Francisca Espinoza
- Neurodevelopmental Biology Unit, Biomedical Sciences Research Laboratory, Basic Sciences Department, Faculty of Medicine, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - Ariel Ávila
- Neurodevelopmental Biology Unit, Biomedical Sciences Research Laboratory, Basic Sciences Department, Faculty of Medicine, Universidad Católica de la Santísima Concepción, Concepción, Chile.
| |
Collapse
|
4
|
Artuyants A, Guo G, Flinterman M, Middleditch M, Jacob B, Lee K, Vella L, Su H, Wilson M, Eva L, Shelling AN, Blenkiron C. The tumour-derived extracellular vesicle proteome varies by endometrial cancer histology and is confounded by an obesogenic environment. Proteomics 2024; 24:e2300055. [PMID: 38644352 DOI: 10.1002/pmic.202300055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/23/2024]
Abstract
Endometrial cancer, the most common gynaecological cancer worldwide, is closely linked to obesity and metabolic diseases, particularly in younger women. New circulating biomarkers have the potential to improve diagnosis and treatment selections, which could significantly improve outcomes. Our approach focuses on extracellular vesicle (EV) biomarker discovery by directly profiling the proteome of EVs enriched from frozen biobanked endometrial tumours. We analysed nine tissue samples to compare three clinical subgroups-low BMI (Body Mass Index) Endometrioid, high BMI Endometrioid, and Serous (any BMI)-identifying proteins related to histological subtype, BMI, and shared secreted proteins. Using collagenase digestion and size exclusion chromatography, we successfully enriched generous quantities of EVs (range 204.8-1291.0 µg protein: 1.38 × 1011-1.10 × 1012 particles), characterised by their size (∼150 nm), expression of EV markers (CD63/81), and proposed endometrial cancer markers (L1CAM, ANXA2). Mass spectrometry-based proteomic profiling identified 2075 proteins present in at least one of the 18 samples. Compared to cell lysates, EVs were successfully depleted for mitochondrial and blood proteins and enriched for common EV markers and large secreted proteins. Further analysis highlighted significant differences in EV protein profiles between the high BMI subgroup and others, underlining the impact of comorbidities on the EV secretome. Interestingly, proteins differentially abundant in tissue subgroups were largely not also differential in matched EVs. This research identified secreted proteins known to be involved in endometrial cancer pathophysiology and proposed novel diagnostic biomarkers (EIF6, MUC16, PROM1, SLC26A2).
Collapse
Affiliation(s)
- Anastasiia Artuyants
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland, New Zealand
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | - George Guo
- Department of Physiology in the School of Medical Sciences, The University of Auckland, Auckland, New Zealand
- Mass Spectrometry Hub, The University of Auckland, Auckland, New Zealand
| | - Marcella Flinterman
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | - Martin Middleditch
- Technical Services, Faculty of Science, The University of Auckland, Auckland, New Zealand
| | - Bincy Jacob
- Centre of eResearch, Faculty of Science, The University of Auckland, Auckland, New Zealand
| | - Kate Lee
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland, New Zealand
| | - Laura Vella
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria, Australia
| | - Huaqi Su
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Michelle Wilson
- Cancer and Blood, Auckland City Hospital, Auckland, New Zealand
- Department of Oncology, The University of Auckland, Auckland, New Zealand
| | - Lois Eva
- Department of Gynaecological Oncology, Auckland City Hospital, Auckland, New Zealand
- Department of Obstetrics and Gynaecology, The University of Auckland, Auckland, New Zealand
| | - Andrew N Shelling
- Department of Obstetrics and Gynaecology, The University of Auckland, Auckland, New Zealand
- Centre for Cancer Research, The University of Auckland, Auckland, New Zealand
| | - Cherie Blenkiron
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland, New Zealand
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
5
|
Lu H, Jing Y, Zhang C, Ma S, Zhang W, Huang D, Zhang B, Zuo Y, Qin Y, Liu GH, Yu Y, Qu J, Wang S. Aging hallmarks of the primate ovary revealed by spatiotemporal transcriptomics. Protein Cell 2024; 15:364-384. [PMID: 38126810 DOI: 10.1093/procel/pwad063] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 10/29/2023] [Indexed: 12/23/2023] Open
Abstract
The ovary is indispensable for female reproduction, and its age-dependent functional decline is the primary cause of infertility. However, the molecular basis of ovarian aging in higher vertebrates remains poorly understood. Herein, we apply spatiotemporal transcriptomics to benchmark architecture organization as well as cellular and molecular determinants in young primate ovaries and compare these to aged primate ovaries. From a global view, somatic cells within the non-follicle region undergo more pronounced transcriptional fluctuation relative to those in the follicle region, likely constituting a hostile microenvironment that facilitates ovarian aging. Further, we uncovered that inflammation, the senescent-associated secretory phenotype, senescence, and fibrosis are the likely primary contributors to ovarian aging (PCOA). Of note, we identified spatial co-localization between a PCOA-featured spot and an unappreciated MT2 (Metallothionein 2) highly expressing spot (MT2high) characterized by high levels of inflammation, potentially serving as an aging hotspot in the primate ovary. Moreover, with advanced age, a subpopulation of MT2high accumulates, likely disseminating and amplifying the senescent signal outward. Our study establishes the first primate spatiotemporal transcriptomic atlas, advancing our understanding of mechanistic determinants underpinning primate ovarian aging and unraveling potential biomarkers and therapeutic targets for aging and age-associated human ovarian disorders.
Collapse
Affiliation(s)
- Huifen Lu
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Ying Jing
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Chen Zhang
- The Fifth People's Hospital of Chongqing, Chongqing 400062, China
| | - Shuai Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- Institute for Stem cell and Regeneration, CAS, Beijing 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Aging Biomarker Consortium, Beijing 100101, China
| | - Weiqi Zhang
- University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem cell and Regeneration, CAS, Beijing 100101, China
- China National Center for Bioinformation, Beijing 100101, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing 101408, China
- Sino-Danish Center for Education and Research, Beijing 101408, China
- Aging Biomarker Consortium, Beijing 100101, China
| | - Daoyuan Huang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Bin Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuesheng Zuo
- University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- China National Center for Bioinformation, Beijing 100101, China
| | - Yingying Qin
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Clinical Research Center for Reproductive Health, Jinan 250012, China
| | - Guang-Hui Liu
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- Institute for Stem cell and Regeneration, CAS, Beijing 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Aging Biomarker Consortium, Beijing 100101, China
| | - Yang Yu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University, Third Hospital, Beijing 100191, China
- Clinical Stem Cell Research Center, Peking University, Third Hospital, Beijing 100191, China
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- Institute for Stem cell and Regeneration, CAS, Beijing 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Aging Biomarker Consortium, Beijing 100101, China
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- The Fifth People's Hospital of Chongqing, Chongqing 400062, China
- Aging Biomarker Consortium, Beijing 100101, China
| |
Collapse
|
6
|
Taylor J, Uhl L, Moll I, Hasan SS, Wiedmann L, Morgenstern J, Giaimo BD, Friedrich T, Alsina-Sanchis E, De Angelis Rigotti F, Mülfarth R, Kaltenbach S, Schenk D, Nickel F, Fleming T, Sprinzak D, Mogler C, Korff T, Billeter AT, Müller-Stich BP, Berriel Diaz M, Borggrefe T, Herzig S, Rohm M, Rodriguez-Vita J, Fischer A. Endothelial Notch1 signaling in white adipose tissue promotes cancer cachexia. NATURE CANCER 2023; 4:1544-1560. [PMID: 37749321 PMCID: PMC10663158 DOI: 10.1038/s43018-023-00622-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/20/2023] [Indexed: 09/27/2023]
Abstract
Cachexia is a major cause of morbidity and mortality in individuals with cancer and is characterized by weight loss due to adipose and muscle tissue wasting. Hallmarks of white adipose tissue (WAT) remodeling, which often precedes weight loss, are impaired lipid storage, inflammation and eventually fibrosis. Tissue wasting occurs in response to tumor-secreted factors. Considering that the continuous endothelium in WAT is the first line of contact with circulating factors, we postulated whether the endothelium itself may orchestrate tissue remodeling. Here, we show using human and mouse cancer models that during precachexia, tumors overactivate Notch1 signaling in distant WAT endothelium. Sustained endothelial Notch1 signaling induces a WAT wasting phenotype in male mice through excessive retinoic acid production. Pharmacological blockade of retinoic acid signaling was sufficient to inhibit WAT wasting in a mouse cancer cachexia model. This demonstrates that cancer manipulates the endothelium at distant sites to mediate WAT wasting by altering angiocrine signals.
Collapse
Affiliation(s)
- Jacqueline Taylor
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Leonie Uhl
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Iris Moll
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sana Safatul Hasan
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Lena Wiedmann
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jakob Morgenstern
- Department of Internal Medicine Endocrinology and Clinical Chemistry, University of Heidelberg, Heidelberg, Germany
| | | | - Tobias Friedrich
- Institute of Biochemistry, University of Giessen, Giessen, Germany
- Biomedical Informatics and Systems Medicine, Science Unit for Basic and Clinical Medicine, Giessen, Germany
| | - Elisenda Alsina-Sanchis
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Francesca De Angelis Rigotti
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Tumor-Stroma Communication Laboratory, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Ronja Mülfarth
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sarah Kaltenbach
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Darius Schenk
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Felix Nickel
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Thomas Fleming
- Department of Internal Medicine Endocrinology and Clinical Chemistry, University of Heidelberg, Heidelberg, Germany
- German Center of Diabetes Research (DZD), Neuherberg, Germany
| | - David Sprinzak
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Carolin Mogler
- Institute of Pathology, Technical University of Munich School of Medicine, Technical University of Munich, Munich, Germany
| | - Thomas Korff
- Institute of Physiology and Pathophysiology, Department of Cardiovascular Physiology, University of Heidelberg, Heidelberg, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Adrian T Billeter
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Beat P Müller-Stich
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Mauricio Berriel Diaz
- Institute for Diabetes and Cancer, Helmholtz Center Munich, German Center for Diabetes Research (DZD), Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Unit, Department of Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany
| | - Tilman Borggrefe
- Institute of Biochemistry, University of Giessen, Giessen, Germany
| | - Stephan Herzig
- Institute for Diabetes and Cancer, Helmholtz Center Munich, German Center for Diabetes Research (DZD), Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Unit, Department of Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany
- Chair Molecular Metabolic Control, Technical University of Munich, Munich, Germany
| | - Maria Rohm
- Institute for Diabetes and Cancer, Helmholtz Center Munich, German Center for Diabetes Research (DZD), Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Unit, Department of Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany
| | - Juan Rodriguez-Vita
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Tumor-Stroma Communication Laboratory, Centro de Investigación Príncipe Felipe, Valencia, Spain.
| | - Andreas Fischer
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany.
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany.
| |
Collapse
|
7
|
4-Methylumbelliferone Targets Revealed by Public Data Analysis and Liver Transcriptome Sequencing. Int J Mol Sci 2023; 24:ijms24032129. [PMID: 36768453 PMCID: PMC9917189 DOI: 10.3390/ijms24032129] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 01/09/2023] [Accepted: 01/18/2023] [Indexed: 01/25/2023] Open
Abstract
4-methylumbelliferone (4MU) is a well-known hyaluronic acid synthesis inhibitor and an approved drug for the treatment of cholestasis. In animal models, 4MU decreases inflammation, reduces fibrosis, and lowers body weight, serum cholesterol, and insulin resistance. It also inhibits tumor progression and metastasis. The broad spectrum of effects suggests multiple and yet unknown targets of 4MU. Aiming at 4MU target deconvolution, we have analyzed publicly available data bases, including: 1. Small molecule library Bio Assay screening (PubChemBioAssay); 2. GO pathway databases screening; 3. Protein Atlas Database. We also performed comparative liver transcriptome analysis of mice on normal diet and mice fed with 4MU for two weeks. Potential targets of 4MU public data base analysis fall into two big groups, enzymes and transcription factors (TFs), including 13 members of the nuclear receptor superfamily regulating lipid and carbohydrate metabolism. Transcriptome analysis revealed changes in the expression of genes involved in bile acid metabolism, gluconeogenesis, and immune response. It was found that 4MU feeding decreased the accumulation of the glycogen granules in the liver. Thus, 4MU has multiple targets and can regulate cell metabolism by modulating signaling via nuclear receptors.
Collapse
|
8
|
Coronel J, Yu J, Pilli N, Kane MA, Amengual J. The conversion of β-carotene to vitamin A in adipocytes drives the anti-obesogenic effects of β-carotene in mice. Mol Metab 2022; 66:101640. [PMID: 36400405 PMCID: PMC9707038 DOI: 10.1016/j.molmet.2022.101640] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/30/2022] [Accepted: 11/09/2022] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE The β-carotene oxygenase 1 (BCO1) is the enzyme responsible for the cleavage of β-carotene to retinal, the first intermediate in vitamin A formation. Preclinical studies suggest that BCO1 expression is required for dietary β-carotene to affect lipid metabolism. The goal of this study was to generate a gene therapy strategy that over-expresses BCO1 in the adipose tissue and utilizes the β-carotene stored in adipocytes to produce vitamin A and reduce obesity. METHODS We generated a novel adipose-tissue-specific, adeno-associated vector to over-express BCO1 (AT-AAV-BCO1) in murine adipocytes. We tested this vector using a unique model to achieve β-carotene accumulation in the adipose tissue, in which Bco1-/- mice were fed β-carotene. An AT-AAV over-expressing green fluorescent protein was utilized as control. We evaluated the adequate delivery route and optimized cellular and organ specificity, dosage, and exposure of our vectors. We also employed morphometric analyses to evaluate the effect of BCO1 expression in adiposity, as well as HPLC and mass spectrometry to quantify β-carotene and retinoids in tissues, including retinoic acid. RESULTS AT-AAV-BCO1 infusions in the adipose tissue of the mice resulted in the production of retinoic acid, a vitamin A metabolite with strong effects on gene regulation. AT-AAV-BCO1 treatment also reduced adipose tissue size and adipocyte area by 35% and 30%, respectively. These effects were sex-specific, highlighting the complexity of vitamin A metabolism in mammals. CONCLUSIONS The over-expression of BCO1 through delivery of an AT-AAV-BCO1 leads to the conversion of β-carotene to vitamin A in adipocytes, which subsequently results in reduction of adiposity. These studies highlight for the first time the potential of adipose tissue β-carotene as a target for BCO1 over-expression in the reduction of obesity.
Collapse
Affiliation(s)
- Johana Coronel
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Jianshi Yu
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, MD, USA
| | - Nageswara Pilli
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, MD, USA
| | - Maureen A. Kane
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, MD, USA
| | - Jaume Amengual
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, USA,Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA,Corresponding author. Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
9
|
Choudhuri R, Sowers AL, Chandramouli GVR, Gamson J, Krishna MC, Mitchell JB, Cook JA. The antioxidant tempol transforms gut microbiome to resist obesity in female C3H mice fed a high fat diet. Free Radic Biol Med 2022; 178:380-390. [PMID: 34883252 PMCID: PMC8753776 DOI: 10.1016/j.freeradbiomed.2021.12.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 12/20/2022]
Abstract
The nitroxide, Tempol, prevents obesity related changes in mice fed a high fat diet (HFD). The purpose of this study was to gain insight into the mechanisms that result in such changes by Tempol in female C3H mice. Microarray methodology, Western blotting, bile acid analyses, and gut microbiome sequencing were used to identify multiple genes, proteins, bile acids, and bacteria that are regulated by Tempol in female C3H mice on HFD. The effects of antibiotics in combination with Tempol on the gut microflora were also studied. Adipose tissue, from Tempol treated mice, was analyzed using targeted gene microarrays revealing up-regulation of fatty acid metabolism genes (Acadm and Acadl > 4-fold, and Acsm3 and Acsm5 > 10-fold). Gene microarray studies of liver tissue from mice switched from HFD to Tempol HFD showed down-regulation of fatty acid synthesis genes and up-regulation of fatty acid oxidation genes. Analyses of proteins involved in obesity revealed that the expression of aldehyde dehydrogenase 1A1 (ALDH1A1) and fasting induced adipose factor/angiopoietin-like protein 4 (FIAF/ANGPTL4) was altered by Tempol HFD. Bile acid studies revealed increases in cholic acid (CA) and deoxycholic acid (DCA) in both the liver and serum of Tempol treated mice. Tempol HFD effect on the gut microbiome composition showed an increase in the population of Akkermansia muciniphila, a bacterial species known to be associated with a lean, anti-inflammatory phenotype. Antibiotic treatment significantly reduced the total level of bacterial numbers, however, Tempol was still effective in reducing the HFD weight gain. Even after antibiotic treatment Tempol still positively influenced several bacterial species such as as Akkermansia muciniphila and Bilophila wadsworthia. The positive effects of Tempol moderating weight gain in female mice fed a HFD involves changes to the gut microbiome, bile acids composition, and finally to changes in genes and proteins involved in fatty acid metabolism and storage.
Collapse
Affiliation(s)
- Rajani Choudhuri
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Anastasia L Sowers
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | - Janet Gamson
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Murali C Krishna
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - James B Mitchell
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - John A Cook
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
10
|
Borel P, Troadec R, Damiani M, Halimi C, Nowicki M, Guichard P, Margier M, Astier J, Grino M, Reboul E, Landrier JF. β-Carotene Bioavailability and Conversion Efficiency Are Significantly Affected by Sex in Rats: First Observation Suggesting a Possible Hormetic Regulation of Vitamin A Metabolism in Female Rats. Mol Nutr Food Res 2021; 65:e2100650. [PMID: 34633772 DOI: 10.1002/mnfr.202100650] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/07/2021] [Indexed: 12/15/2022]
Abstract
SCOPE To study the effect of variation in dietary vitamin A (VA) content on its hepatic and intestinal metabolism. METHODS AND RESULTS Adult female and male rats are fed with diets containing 400, 2300, or 9858 IU kg-1 VA for 31-33 weeks. VA concentrations are measured in plasma and liver. Bioavailability and intestinal conversion efficiency of β-carotene to VA are assessed by measuring postprandial plasma β-carotene and retinyl palmitate concentrations after force-feeding rats with β-carotene. Expression of genes involved in VA metabolism, together with concentrations of RBP4, BCO1, and SR-BI proteins, are measured in the intestine and liver of female rats. Plasma retinol concentrations are lower and hepatic free retinol concentrations are higher in females than in males. There is no effect of dietary VA content on β-carotene bioavailability and its conversion efficiency, but bioavailability is higher and conversion efficiency is lower in females than in males. The expression of most genes exhibited a U-shaped dose response curve depending on VA intake. CONCLUSIONS β-Carotene bioavailability and conversion efficiency to VA are affected by the sex of rats. Results of gene expression suggest a hormetic regulation of VA metabolism in female rats.
Collapse
Affiliation(s)
- Patrick Borel
- C2VN, INRAE, INSERM, Aix-Marseille Univ, Marseille, France
| | - Romane Troadec
- C2VN, INRAE, INSERM, Aix-Marseille Univ, Marseille, France
| | | | | | - Marion Nowicki
- C2VN, INRAE, INSERM, Aix-Marseille Univ, Marseille, France
| | | | | | - Julien Astier
- C2VN, INRAE, INSERM, Aix-Marseille Univ, Marseille, France
| | - Michel Grino
- C2VN, INRAE, INSERM, Aix-Marseille Univ, Marseille, France
| | | | | |
Collapse
|
11
|
Khatun M, Meltsov A, Lavogina D, Loid M, Kask K, Arffman RK, Rossi HR, Lättekivi F, Jääger K, Krjutškov K, Rinken A, Salumets A, Piltonen TT. Decidualized endometrial stromal cells present with altered androgen response in PCOS. Sci Rep 2021; 11:16287. [PMID: 34381107 PMCID: PMC8357821 DOI: 10.1038/s41598-021-95705-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 07/23/2021] [Indexed: 01/27/2023] Open
Abstract
Hyperandrogenic women with PCOS show disrupted decidualization (DE) and placentation. Dihydrotestosterone (DHT) is reported to enhance DE in non-PCOS endometrial stromal cells (eSCCtrl); however, this has not been assessed in PCOS cells (eSCPCOS). Therefore, we studied the transcriptome profile of non-decidualized (non-DE) and DE eSCs from women with PCOS and Ctrl in response to short-term estradiol (E2) and/or progesterone (P4) exposure with/without (±) DHT. The non-DE eSCs were subjected to E2 ± DHT treatment, whereas the DE (0.5 mM 8-Br-cAMP, 96 h) eSCs were post-treated with E2 and P4 ± DHT, and RNA-sequenced. Validation was performed by immunofluorescence and immunohistochemistry. The results showed that, regardless of treatment, the PCOS and Ctrl samples clustered separately. The comparison of DE vs. non-DE eSCPCOS without DHT revealed PCOS-specific differentially expressed genes (DEGs) involved in mitochondrial function and progesterone signaling. When further adding DHT, we detected altered responses for lysophosphatidic acid (LPA), inflammation, and androgen signaling. Overall, the results highlight an underlying defect in decidualized eSCPCOS, present with or without DHT exposure, and possibly linked to the altered pregnancy outcomes. We also report novel factors which elucidate the mechanisms of endometrial dysfunction in PCOS.
Collapse
Affiliation(s)
- Masuma Khatun
- Department of Obstetrics and Gynaecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Alvin Meltsov
- Competence Centre on Health Technologies, Tartu, Estonia.,Department of Computer Science, University of Tartu, Tartu, Estonia
| | - Darja Lavogina
- Competence Centre on Health Technologies, Tartu, Estonia.,Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Marina Loid
- Competence Centre on Health Technologies, Tartu, Estonia.,Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Keiu Kask
- Competence Centre on Health Technologies, Tartu, Estonia.,Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Riikka K Arffman
- Department of Obstetrics and Gynaecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Henna-Riikka Rossi
- Department of Obstetrics and Gynaecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Freddy Lättekivi
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia.,Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Tartu, Estonia
| | - Kersti Jääger
- Competence Centre on Health Technologies, Tartu, Estonia
| | - Kaarel Krjutškov
- Competence Centre on Health Technologies, Tartu, Estonia.,Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Ago Rinken
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Andres Salumets
- Competence Centre on Health Technologies, Tartu, Estonia.,Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia.,Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Terhi T Piltonen
- Department of Obstetrics and Gynaecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland.
| |
Collapse
|
12
|
Abstract
Dietary intake and tissue levels of carotenoids have been associated with a reduced risk of several chronic diseases, including cardiovascular diseases, type 2 diabetes, obesity, brain-related diseases and some types of cancer. However, intervention trials with isolated carotenoid supplements have mostly failed to confirm the postulated health benefits. It has thereby been speculated that dosing, matrix and synergistic effects, as well as underlying health and the individual nutritional status plus genetic background do play a role. It appears that our knowledge on carotenoid-mediated health benefits may still be incomplete, as the underlying mechanisms of action are poorly understood in relation to human relevance. Antioxidant mechanisms - direct or via transcription factors such as NRF2 and NF-κB - and activation of nuclear hormone receptor pathways such as of RAR, RXR or also PPARs, via carotenoid metabolites, are the basic principles which we try to connect with carotenoid-transmitted health benefits as exemplified with described common diseases including obesity/diabetes and cancer. Depending on the targeted diseases, single or multiple mechanisms of actions may play a role. In this review and position paper, we try to highlight our present knowledge on carotenoid metabolism and mechanisms translatable into health benefits related to several chronic diseases.
Collapse
|
13
|
Petrosino JM, Longenecker JZ, Ramkumar S, Xu X, Dorn LE, Bratasz A, Yu L, Maurya S, Tolstikov V, Bussberg V, Janssen PM, Periasamy M, Kiebish MA, Duester G, von Lintig J, Ziouzenkova O, Accornero F. Paracardial fat remodeling affects systemic metabolism through alcohol dehydrogenase 1. J Clin Invest 2021; 131:141799. [PMID: 33586683 PMCID: PMC7880313 DOI: 10.1172/jci141799] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 12/10/2020] [Indexed: 11/17/2022] Open
Abstract
The relationship between adiposity and metabolic health is well established. However, very little is known about the fat depot, known as paracardial fat (pCF), located superior to and surrounding the heart. Here, we show that pCF remodels with aging and a high-fat diet and that the size and function of this depot are controlled by alcohol dehydrogenase 1 (ADH1), an enzyme that oxidizes retinol into retinaldehyde. Elderly individuals and individuals with obesity have low ADH1 expression in pCF, and in mice, genetic ablation of Adh1 is sufficient to drive pCF accumulation, dysfunction, and global impairments in metabolic flexibility. Metabolomics analysis revealed that pCF controlled the levels of circulating metabolites affecting fatty acid biosynthesis. Also, surgical removal of the pCF depot was sufficient to rescue the impairments in cardiometabolic flexibility and fitness observed in Adh1-deficient mice. Furthermore, treatment with retinaldehyde prevented pCF remodeling in these animals. Mechanistically, we found that the ADH1/retinaldehyde pathway works by driving PGC-1α nuclear translocation and promoting mitochondrial fusion and biogenesis in the pCF depot. Together, these data demonstrate that pCF is a critical regulator of cardiometabolic fitness and that retinaldehyde and its generating enzyme ADH1 act as critical regulators of adipocyte remodeling in the pCF depot.
Collapse
Affiliation(s)
- Jennifer M. Petrosino
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Jacob Z. Longenecker
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | | | - Xianyao Xu
- Department of Biomedical Engineering, Dorothy M. Davis Heart and Lung Research Institute
| | - Lisa E. Dorn
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | | | - Lianbo Yu
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio, USA
| | - Santosh Maurya
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | | | - Valerie Bussberg
- BERG, Precision Medicine Department, Framingham, Massachusetts, USA
| | - Paul M.L. Janssen
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Muthu Periasamy
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
- Department of Internal Medicine, University of Central Florida, Orlando, Florida, USA
| | | | - Gregg Duester
- Development, Aging, and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Johannes von Lintig
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Ouliana Ziouzenkova
- Department of Human Sciences, College of Education and Human Ecology, Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Federica Accornero
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
14
|
L-carnitine exerts a nutrigenomic effect via direct modulation of nuclear receptor signaling in adipocytes, hepatocytes and SKMC, demonstrating its nutritional impact. Nutr Res 2020; 85:84-98. [PMID: 33453499 DOI: 10.1016/j.nutres.2020.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/13/2020] [Accepted: 11/10/2020] [Indexed: 12/13/2022]
Abstract
L-carnitine is an indispensable metabolite facilitating the transport of fatty acids into the mitochondrial matrix and has been previously postulated to exert a nutrigenomic effect. However, the underlying molecular mechanisms remain mostly unclear. We hypothesized that L-carnitine interacts with nuclear receptors involved in metabolic regulation, thereby modulating downstream targets of cellular metabolism. Therefore, we investigated the effect of L-carnitine supplementation on protein activity, mRNA expression, and binding affinities of nuclear receptors as well as mRNA expression of downstream targets in skeletal muscle cells, hepatocytes, and differentiated adipocytes. L-carnitine supplementation to hepatocytes increased the protein activity of multiple nuclear receptors (RAR, RXR, VDR, PPAR, HNF4, ER, LXR). Diverging effects on the mRNA expression of PPAR-α, PPAR-δ, PPAR-γ, RAR-β, LXR-α, and RXR-α were observed in adipocytes, hepatocytes, and skeletal muscle cells. mRNA levels of PPAR-α, a key regulator of lipolysis and β-oxidation, were significantly upregulated, emphasizing a role of L-carnitine as a promoter of lipid catabolism. L-carnitine administration to hepatocytes modulated the transcription of key nuclear receptor target genes, including ALDH1A1, a promoter of adipogenesis, and OGT, a contributor to insulin resistance. Electrophoretic mobility shift assays proved L-carnitine to increase binding affinities of nuclear receptors to their promoter target sequences, suggesting a molecular mechanism for the observed transcriptional modulation. Overall, these findings indicate that L-carnitine modulates the activity and expression of nuclear receptors, thereby promoting lipolytic gene expression and decreasing transcription of target genes linked to adipogenesis and insulin resistance.
Collapse
|
15
|
Hand osteoarthritis: clinical phenotypes, molecular mechanisms and disease management. Nat Rev Rheumatol 2019; 14:641-656. [PMID: 30305701 DOI: 10.1038/s41584-018-0095-4] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Osteoarthritis (OA) is a highly prevalent condition, and the hand is the most commonly affected site. Patients with hand OA frequently report symptoms of pain, functional limitations and frustration in undertaking everyday activities. The condition presents clinically with changes to the bone, ligaments, cartilage and synovial tissue, which can be observed using radiography, ultrasonography or MRI. Hand OA is a heterogeneous disorder and is considered to be multifactorial in aetiology. This Review provides an overview of the epidemiology, presentation and burden of hand OA, including an update on hand OA imaging (including the development of novel techniques), disease mechanisms and management. In particular, areas for which new evidence has substantially changed the way we understand, consider and treat hand OA are highlighted. For example, genetic studies, clinical trials and careful prospective imaging studies from the past 5 years are beginning to provide insights into the pathogenesis of hand OA that might uncover new therapeutic targets in the disease.
Collapse
|
16
|
Farmahin R, Gannon AM, Gagné R, Rowan-Carroll A, Kuo B, Williams A, Curran I, Yauk CL. Hepatic transcriptional dose-response analysis of male and female Fischer rats exposed to hexabromocyclododecane. Food Chem Toxicol 2018; 133:110262. [PMID: 30594549 DOI: 10.1016/j.fct.2018.12.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 12/13/2018] [Accepted: 12/20/2018] [Indexed: 12/14/2022]
Abstract
Hexabromocyclododecane (HBCD) is a brominated flame retardant found in the environment and human tissues. The toxicological effects of HBCD exposure are not clearly understood. We employed whole-genome RNA-sequencing on liver samples from male and female Fischer rats exposed to 0, 250, 1250, and 5000 mg technical mixture of HBCD/kg diet for 28 days to gain further insight into HBCD toxicity. HBCD altered 428 and 250 gene transcripts in males and females, respectively, which were involved in metabolism of xenobiotics, oxidative stress, immune response, metabolism of glucose and lipids, circadian regulation, cell cycle, fibrotic activity, and hormonal balance. Signature analysis supported that HBCD operates through the constitutive androstane and pregnane X receptors. The median transcriptomic benchmark dose (BMD) for the lowest statistically significant pathway was within 1.5-fold of the BMD for increased liver weight, while the BMD for the lowest pathway with at least three modeled genes (minimum 5% of pathway) was similar to the lowest apical endpoint BMD. The results show how transcriptional analyses can inform mechanisms underlying chemical toxicity and the doses at which potentially adverse effects occur. This experiment is part of a larger study exploring the use of toxicogenomics and high-throughput screening for human health risk assessment.
Collapse
Affiliation(s)
- Reza Farmahin
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Anne Marie Gannon
- Regulatory Toxicology Research Division, Health Products and Food Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Rémi Gagné
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Andrea Rowan-Carroll
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Byron Kuo
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Andrew Williams
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Ivan Curran
- Regulatory Toxicology Research Division, Health Products and Food Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Carole L Yauk
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada.
| |
Collapse
|
17
|
Design, synthesis, and ex vivo evaluation of a selective inhibitor for retinaldehyde dehydrogenase enzymes. Bioorg Med Chem 2018; 26:5766-5779. [PMID: 30409702 DOI: 10.1016/j.bmc.2018.10.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 10/03/2018] [Accepted: 10/12/2018] [Indexed: 11/22/2022]
Abstract
The retinaldehyde dehydrogenase (RALDH) enzymes, RALDH1, RALDH2, and RALDH3, catalyze the irreversible oxidation of retinaldehyde to all-trans-retinoic acid (ATRA). Despite the importance of the RALDH enzymes in embryonic development, postnatal growth and differentiation, and in several disease states, there are no commercially available inhibitors that specifically target these isozymes. We report here the development and characterization of a small molecule inhibitor dichloro-all-trans-retinone (DAR) (Summers et al., 2017) that is an irreversible inhibitor of RALDH1, 2, and 3 that effectively inhibits RALDH1, 2, and 3 in the nanomolar range but has no inhibitory activity against mitochondrial ALDH2. These results provide support for the development of DAR as a specific ATRA synthesis inhibitor for a variety of experimental and clinical applications.
Collapse
|
18
|
Takitani K, Kishi K, Miyazaki H, Koh M, Tamaki H, Inoue A, Tamai H. Altered Expression of Retinol Metabolism-Related Genes in an ANIT-Induced Cholestasis Rat Model. Int J Mol Sci 2018; 19:ijms19113337. [PMID: 30373117 PMCID: PMC6274878 DOI: 10.3390/ijms19113337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 10/19/2018] [Accepted: 10/23/2018] [Indexed: 11/16/2022] Open
Abstract
Cholestasis is defined as a reduction of bile secretion caused by a dysfunction of bile formation. Insufficient bile secretion into the intestine undermines the formation of micelles, which may result in the reduced absorption of lipids and fat-soluble vitamins. Here, we investigated the retinol homeostasis and the alterations of retinol metabolism-related genes, including β-carotene 15,15′ monooxygenase (BCMO), lecithin:retinol acyltransferase (LRAT), aldehyde dehydrogenase (ALDH), cytochrome P450 26A1 (CYP26A1), and retinoic acid receptors (RAR) β, in a α-naphthyl isothiocyanate (ANIT)-induced cholestasis rat model. Moreover, we examined the expression of the farnesoid X receptor (FXR) target genes. Our results showed that plasma retinol levels were decreased in ANIT rats compared to control rats. On the contrary, hepatic retinol levels were not different between the two groups. The expression of FXR target genes in the liver and intestine of cholestasis model rats was repressed. The BCMO expression was decreased in the liver and increased in the intestine of ANIT rats compared to control rats. Finally, the hepatic expression of LRAT, RARβ, and ALDH1A1 in cholestatic rats was decreased compared to the control rats, while the CYP26A1 expression of the liver was not altered. The increased expression of intestinal BCMO in cholestasis model rats might compensate for decreased circulatory retinol levels. The BCMO expression might be regulated in a tissue-specific manner to maintain the homeostasis of retinol.
Collapse
Affiliation(s)
- Kimitaka Takitani
- Department of Pediatrics, Osaka Medical College, Osaka 569-8686, Japan.
| | - Kanta Kishi
- Department of Pediatrics, Osaka Medical College, Osaka 569-8686, Japan.
| | - Hiroshi Miyazaki
- Department of Pediatrics, Osaka Medical College, Osaka 569-8686, Japan.
- Department of Pediatrics, Osaka Rosai Hospital, Osaka 591-8025, Japan.
| | - Maki Koh
- Department of Pediatrics, Osaka Medical College, Osaka 569-8686, Japan.
| | - Hirofumi Tamaki
- Department of Pediatrics, Osaka Medical College, Osaka 569-8686, Japan.
- Department of Medicine, Shinseikai Daiichi Hospital, Aichi 468-0031, Japan.
| | - Akiko Inoue
- Department of Pediatrics, Osaka Medical College, Osaka 569-8686, Japan.
| | - Hiroshi Tamai
- Department of Pediatrics, Osaka Medical College, Osaka 569-8686, Japan.
| |
Collapse
|
19
|
Vitamin A Deficiency and the Lung. Nutrients 2018; 10:nu10091132. [PMID: 30134568 PMCID: PMC6164133 DOI: 10.3390/nu10091132] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 08/13/2018] [Accepted: 08/17/2018] [Indexed: 12/12/2022] Open
Abstract
Vitamin A (all-trans-retinol) is a fat-soluble micronutrient which together with its natural derivatives and synthetic analogues constitutes the group of retinoids. They are involved in a wide range of physiological processes such as embryonic development, vision, immunity and cellular differentiation and proliferation. Retinoic acid (RA) is the main active form of vitamin A and multiple genes respond to RA signalling through transcriptional and non-transcriptional mechanisms. Vitamin A deficiency (VAD) is a remarkable public health problem. An adequate vitamin A intake is required in early lung development, alveolar formation, tissue maintenance and regeneration. In fact, chronic VAD has been associated with histopathological changes in the pulmonary epithelial lining that disrupt the normal lung physiology predisposing to severe tissue dysfunction and respiratory diseases. In addition, there are important alterations of the structure and composition of extracellular matrix with thickening of the alveolar basement membrane and ectopic deposition of collagen I. In this review, we show our recent findings on the modification of cell-junction proteins in VAD lungs, summarize up-to-date information related to the effects of chronic VAD in the impairment of lung physiology and pulmonary disease which represent a major global health problem and provide an overview of possible pathways involved.
Collapse
|
20
|
Kozovska Z, Patsalias A, Bajzik V, Durinikova E, Demkova L, Jargasova S, Smolkova B, Plava J, Kucerova L, Matuskova M. ALDH1A inhibition sensitizes colon cancer cells to chemotherapy. BMC Cancer 2018; 18:656. [PMID: 29902974 PMCID: PMC6003038 DOI: 10.1186/s12885-018-4572-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 05/31/2018] [Indexed: 12/18/2022] Open
Abstract
Background Recent evidence in cancer research, developed the notion that malignant tumors consist of different subpopulations of cells, one of them, known as cancer stem cells, being attributed many important properties such as enhanced tumorigenicity, proliferation potential and profound multidrug resistance to chemotherapy. Several key stem cells markers were identified in colon cancer. In our study we focused on the aldehyde dehydrogenase type 1 (ALDH1) expression in colon cancer-derived cell lines HT-29/eGFP, HCT-116/eGFP and LS-180/eGFP, and its role in the chemoresistance and tumorigenic potential. Methods The effect of pharmacological inhibition of ALDH activity by diethylaminobenzaldehyde (DEAB) and also effect of molecular inhibition by specific siRNA was evaluated in vitro in cultures of human colorectal cell lines. The expression level of different isoenzymes of aldehyde dehydrogenase was determined using qPCR. Changes in cell biology were evaluated by expression analysis, western blot and apoptosis assay. The efficiency of cytotoxic treatment in the presence of different chemotherapeutic drugs was analyzed by fluorimetric assay. Tumorigenicity of cells with specific ALDH1A1 siRNA was tested in xenograft model in vivo. Results Treatment by DEAB partially sensitized the tested cell lines to chemotherapeutics. Subsequently the molecular inhibition of specific isoforms of ALDH by ALDH1A1 or ALDH1A3 siRNA led to sensitizing of cell lines HT-29/eGFP, HCT-116/eGFP to capecitabine and 5-FU. On the model of athymic mice we observed the effect of molecular inhibition of ALDH1A1 in HT-29/eGFP cells by siRNA. We observed inhibition of proliferation of subcutaneous xenografts in comparison to control cells. Conclusion This research, verifies the significance of the ALDH1A isoforms in multidrug resistance of human colorectal cancer cells and its potential as a cancer stem cell marker. This provides the basis for the development of new approaches regarding the treatment of patients with colorectal adenocarcinoma and potentially the treatment of other tumor malignancies.
Collapse
Affiliation(s)
- Z Kozovska
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of SAS, Dubravska cesta 9, 845 05, Bratislava, Slovakia.
| | - A Patsalias
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of SAS, Dubravska cesta 9, 845 05, Bratislava, Slovakia
| | - V Bajzik
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of SAS, Dubravska cesta 9, 845 05, Bratislava, Slovakia
| | - E Durinikova
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of SAS, Dubravska cesta 9, 845 05, Bratislava, Slovakia
| | - L Demkova
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of SAS, Dubravska cesta 9, 845 05, Bratislava, Slovakia
| | - S Jargasova
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of SAS, Dubravska cesta 9, 845 05, Bratislava, Slovakia
| | - B Smolkova
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of SAS, Dubravska cesta 9, 845 05, Bratislava, Slovakia
| | - J Plava
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of SAS, Dubravska cesta 9, 845 05, Bratislava, Slovakia
| | - L Kucerova
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of SAS, Dubravska cesta 9, 845 05, Bratislava, Slovakia
| | - M Matuskova
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of SAS, Dubravska cesta 9, 845 05, Bratislava, Slovakia
| |
Collapse
|
21
|
Israelian N, Danska JS. Sex Effects at the Ramparts: Nutrient- and Microbe-Mediated Regulation of the Immune-Metabolic Interface. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1043:113-140. [PMID: 29224093 DOI: 10.1007/978-3-319-70178-3_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The relationships between dietary compounds, derivative metabolites, and host metabolism and immunity are controlled by diverse molecular mechanisms. Essential contributions to these dynamics come from the community of microbes (the microbiome) inhabiting the human digestive tract. The composition and function of the microbiome are shaped by available nutrients, and reciprocally, these organisms produce an as yet poorly defined repertoire of molecules that communicate with the epithelial barrier and the mucosal immune system. We present evidence that diet-derived vitamins and lipids regulate immunity and metabolic function and highlight the diverse mechanisms through which these effects are impacted by sex. We discuss exciting new data emerging from studies using high-throughput sequencing technology, specialized mouse models, and bio-specimens, and clinical data from human subjects that have begun to reveal the complexity of these interactions. Also profiled in this chapter are the striking sex differences in pathways by which dietary nutrients and gut microbes modify metabolism, immunity, and immune- and inflammation-mediated diseases. Although the incidence, severity, and therapeutic responses of many autoimmune diseases differ by sex, the molecular mechanisms of these effects remain poorly understood.
Collapse
Affiliation(s)
- Nyrie Israelian
- Department of Immunology, University of Toronto, Toronto, ON, Canada.,Program in Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON, Canada
| | - Jayne S Danska
- Program in Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON, Canada. .,Department of Immunology, and Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
22
|
|
23
|
Pavăl D, Rad F, Rusu R, Niculae AŞ, Colosi HA, Dobrescu I, Dronca E. Low Retinal Dehydrogenase 1 (RALDH1) Level in Prepubertal Boys with Autism Spectrum Disorder: A Possible Link to Dopamine Dysfunction? CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2017; 15:229-236. [PMID: 28783931 PMCID: PMC5565080 DOI: 10.9758/cpn.2017.15.3.229] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/23/2016] [Accepted: 09/05/2016] [Indexed: 11/18/2022]
Abstract
Objective Retinal dehydrogenase 1 (RALDH1) is a cytosolic enzyme which acts both as a source of retinoic acid (RA) and as a detoxification enzyme. RALDH1 has key functions in the midbrain dopaminergic system, which influences motivation, cognition, and social behavior. Since dopamine has been increasingly linked to autism spectrum disorder (ASD), we asked whether RALDH1 could contribute to the autistic phenotype. Therefore, we investigated for the first time the levels of RALDH1 in autistic patients. To further assess the detoxification function of RALDH1, we also explored 4-hydroxynonenal protein adducts (4-HNE PAs) and reduced glutathione (GSH) levels. Moreover, considering the effect of testosterone on RALDH1 expression, we measured the second to fourth digit ratio (2D:4D ratio) for both hands, which reflects exposure to prenatal testosterone. Methods Male patients with ASD (n=18; age, 62.9±4.3 months) and healthy controls (n=13; age, 78.1±4.9 months) were examined. Erythrocyte RALDH1, serum 4-HNE PAs and erythrocyte GSH levels were measured using colorimetric assays, and digit lengths were measured using digital calipers. Results We found significantly lower (−42.9%) RALDH1 levels in autistic patients as compared to controls (p=0.032). However, there was no difference in 4-HNE PAs levels (p=0.368), GSH levels (p=0.586), or 2D:4D ratios (p=0.246 in the left hand, p=0.584 in the right hand) between healthy controls and autistic subjects. Conclusion We concluded that a subset of autistic patients had a low RALDH1 level. These results suggest that low RALDH1 levels could contribute to the autistic phenotype by reflecting a dopaminergic dysfunction.
Collapse
Affiliation(s)
- Denis Pavăl
- Department of Molecular Sciences, Faculty of Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Florina Rad
- Alexandru Obregia Psychiatry Hospital, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Răzvan Rusu
- Department of Molecular Sciences, Faculty of Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Alexandru-Ştefan Niculae
- Department of Molecular Sciences, Faculty of Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Horaţiu Alexandru Colosi
- Department of Medical Informatics and Biostatistics, Faculty of Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Iuliana Dobrescu
- Alexandru Obregia Psychiatry Hospital, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Eleonora Dronca
- Department of Molecular Sciences, Faculty of Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
24
|
Bohn T, Desmarchelier C, Dragsted LO, Nielsen CS, Stahl W, Rühl R, Keijer J, Borel P. Host-related factors explaining interindividual variability of carotenoid bioavailability and tissue concentrations in humans. Mol Nutr Food Res 2017; 61:1600685. [PMID: 28101967 PMCID: PMC5516247 DOI: 10.1002/mnfr.201600685] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 12/19/2016] [Accepted: 01/04/2017] [Indexed: 12/14/2022]
Abstract
Carotenoid dietary intake and their endogenous levels have been associated with a decreased risk of several chronic diseases. There are indications that carotenoid bioavailability depends, in addition to the food matrix, on host factors. These include diseases (e.g. colitis), life-style habits (e.g. smoking), gender and age, as well as genetic variations including single nucleotide polymorphisms that govern carotenoid metabolism. These are expected to explain interindividual differences that contribute to carotenoid uptake, distribution, metabolism and excretion, and therefore possibly also their association with disease risk. For instance, digestion enzymes fostering micellization (PNLIP, CES), expression of uptake/efflux transporters (SR-BI, CD36, NPC1L1), cleavage enzymes (BCO1/2), intracellular transporters (FABP2), secretion into chylomicrons (APOB, MTTP), carotenoid metabolism in the blood and liver (LPL, APO C/E, LDLR), and distribution to target tissues such as adipose tissue or macula (GSTP1, StARD3) depend on the activity of these proteins. In addition, human microbiota, e.g. via altering bile-acid concentrations, may play a role in carotenoid bioavailability. In order to comprehend individual, variable responses to these compounds, an improved knowledge on intra-/interindividual factors determining carotenoid bioavailability, including tissue distribution, is required. Here, we highlight the current knowledge on factors that may explain such intra-/interindividual differences.
Collapse
Affiliation(s)
- Torsten Bohn
- Luxembourg Institute of HealthStrassenLuxembourg
| | | | - Lars O. Dragsted
- Department of Nutrition, Exercise and SportsUniversity of CopenhagenFrederiksberg CDenmark
| | - Charlotte S. Nielsen
- Department of Nutrition, Exercise and SportsUniversity of CopenhagenFrederiksberg CDenmark
| | - Wilhelm Stahl
- Institute of Biochemistry and Molecular Biology IHeinrich‐Heine‐University DüsseldorfDüsseldorfGermany
| | - Ralph Rühl
- Paprika Bioanalytics BTDebrecenHungary
- MTA‐DE Public Health Research Group of the Hungarian Academy of SciencesFaculty of Public HealthUniversity of DebrecenDebrecenHungary
| | - Jaap Keijer
- Human and Animal PhysiologyWageningen UniversityWageningenThe Netherlands
| | - Patrick Borel
- NORT, Aix‐Marseille Université, INRAINSERMMarseilleFrance
| |
Collapse
|
25
|
Braun F, Rinschen MM, Bartels V, Frommolt P, Habermann B, Hoeijmakers JHJ, Schumacher B, Dollé MET, Müller RU, Benzing T, Schermer B, Kurschat CE. Altered lipid metabolism in the aging kidney identified by three layered omic analysis. Aging (Albany NY) 2017; 8:441-57. [PMID: 26886165 PMCID: PMC4833139 DOI: 10.18632/aging.100900] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Aging-associated diseases and their comorbidities affect the life of a constantly growing proportion of the population in developed countries. At the center of these comorbidities are changes of kidney structure and function as age-related chronic kidney disease predisposes to the development of cardiovascular diseases such as stroke, myocardial infarction or heart failure. To detect molecular mechanisms involved in kidney aging, we analyzed gene expression profiles of kidneys from adult and aged wild-type mice by transcriptomic, proteomic and targeted lipidomic methodologies. Interestingly, transcriptome and proteome analyses revealed differential expression of genes primarily involved in lipid metabolism and immune response. Additional lipidomic analyses uncovered significant age-related differences in the total amount of phosphatidylethanolamines, phosphatidylcholines and sphingomyelins as well as in subspecies of phosphatidylserines and ceramides with age. By integration of these datasets we identified Aldh1a1, a key enzyme in vitamin A metabolism specifically expressed in the medullary ascending limb, as one of the most prominent upregulated proteins in old kidneys. Moreover, ceramidase Asah1 was highly expressed in aged kidneys, consistent with a decrease in ceramide C16. In summary, our data suggest that changes in lipid metabolism are involved in the process of kidney aging and in the development of chronic kidney disease.
Collapse
Affiliation(s)
- Fabian Braun
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Markus M Rinschen
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Valerie Bartels
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Department of Cardiology and Angiology, University of Münster, Münster, Germany
| | - Peter Frommolt
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany
| | - Bianca Habermann
- Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany.,Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Jan H J Hoeijmakers
- Department of Cell Biology and Genetics, Medical Genetics Centre, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Björn Schumacher
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Institute for Genome Stability in Aging and Disease, Medical Faculty, University of Cologne, Cologne, Germany
| | - Martijn E T Dollé
- National Institute of Public Health and the Environment, Centre for Health Protection, Bilthoven, The Netherlands
| | - Roman-Ulrich Müller
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany
| | - Bernhard Schermer
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany
| | - Christine E Kurschat
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| |
Collapse
|
26
|
Landrier JF, Kasiri E, Karkeni E, Mihály J, Béke G, Weiss K, Lucas R, Aydemir G, Salles J, Walrand S, de Lera AR, Rühl R. Reduced adiponectin expression after high-fat diet is associated with selective up-regulation of ALDH1A1 and further retinoic acid receptor signaling in adipose tissue. FASEB J 2016; 31:203-211. [PMID: 27729412 PMCID: PMC5161515 DOI: 10.1096/fj.201600263rr] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 09/22/2016] [Indexed: 12/22/2022]
Abstract
Adiponectin is an adipocyte-derived adipokine with potent antidiabetic, anti-inflammatory, and antiatherogenic activity. Long-term, high-fat diet results in gain of body weight, adiposity, further inflammatory-based cardiovascular diseases, and reduced adiponectin secretion. Vitamin A derivatives/retinoids are involved in several of these processes, which mainly take place in white adipose tissue (WAT). In this study, we examined adiponectin expression as a function of dietary high-fat and high–vitamin A conditions in mice. A decrease of adiponectin expression in addition to an up-regulation of aldehyde dehydrogenase A1 (ALDH1A1), retinoid signaling, and retinoic acid response element signaling was selectively observed in WAT of mice fed a normal–vitamin A, high-fat diet. Reduced adiponectin expression in WAT was also observed in mice fed a high–vitamin A diet. Adipocyte cell culture revealed that endogenous and synthetic retinoic acid receptor (RAR)α- and RARγ-selective agonists, as well as a synthetic retinoid X receptor agonist, efficiently reduced adiponectin expression, whereas ALDH1A1 expression only increased with RAR agonists. We conclude that reduced adiponectin expression under high-fat dietary conditions is dependent on 1) increased ALDH1A1 expression in adipocytes, which does not increase all-trans-retinoic acid levels; 2) further RAR ligand–induced, WAT-selective, increased retinoic acid response element–mediated signaling; and 3) RAR ligand–dependent reduction of adiponectin expression.—Landrier, J.-F., Kasiri, E., Karkeni, E., Mihály, J., Béke, G., Weiss, K., Lucas, R., Aydemir, G., Salles, J., Walrand, S., de Lera, A. R., Rühl, R. Reduced adiponectin expression after high-fat diet is associated with selective up-regulation of ALDH1A1 and further retinoic acid receptor signaling in adipose tissue.
Collapse
Affiliation(s)
- Jean-Francois Landrier
- Institut National de la Recherche Agronomique, Unités Mixtes de Recherche 1260, Marseille, France.,INSERM, Unités Mixtes de Recherche 1062, Nutrition, Obésité et Risque Thrombotique, Marseille, France.,Aix-Marseille Université, Faculté de Médecine, Marseille, France
| | - Elnaz Kasiri
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, Hungary.,MTA-DE Public Health Research Group, Hungarian Academy of Sciences, Faculty of Public Health, University of Debrecen, Debrecen, Hungary
| | - Esma Karkeni
- Institut National de la Recherche Agronomique, Unités Mixtes de Recherche 1260, Marseille, France.,INSERM, Unités Mixtes de Recherche 1062, Nutrition, Obésité et Risque Thrombotique, Marseille, France.,Aix-Marseille Université, Faculté de Médecine, Marseille, France
| | - Johanna Mihály
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, Hungary
| | - Gabriella Béke
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, Hungary
| | - Kathrin Weiss
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, Hungary
| | - Renata Lucas
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, Hungary
| | - Gamze Aydemir
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, Hungary
| | - Jérome Salles
- Unités Mixtes de Recherche, Institut National de la Recherche Agronomique (INRA) 1019 Unité de Nutrition Humaine, Centre de Recherches INRA de Clermont-Ferrand/Theix, Saint-Genès-Champanelle, France
| | - Stéphane Walrand
- Unités Mixtes de Recherche, Institut National de la Recherche Agronomique (INRA) 1019 Unité de Nutrition Humaine, Centre de Recherches INRA de Clermont-Ferrand/Theix, Saint-Genès-Champanelle, France
| | - Angel R de Lera
- Departamento de Química Orgánica, Universidade de Vigo, Facultad de Química, Centro de Investigaciones Biomédicas and Instituto de Investigación Biomédica de Vigo, Vigo, Spain; and
| | - Ralph Rühl
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, Hungary; .,MTA-DE Public Health Research Group, Hungarian Academy of Sciences, Faculty of Public Health, University of Debrecen, Debrecen, Hungary.,Paprika Bioanalytics BT, Debrecen, Hungary
| |
Collapse
|
27
|
Human articular chondrocytes with higher aldehyde dehydrogenase activity have stronger expression of COL2A1 and SOX9. Osteoarthritis Cartilage 2016; 24:873-82. [PMID: 26687820 DOI: 10.1016/j.joca.2015.11.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 10/17/2015] [Accepted: 11/24/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To determine in human articular chondrocytes the activity of Aldehyde dehydrogenase (ALDH), which are reported as stem/progenitor cell marker in various adult tissues and evaluate gene expression of ALDH1A isoforms. DESIGN ALDH activity was evaluated by flow cytometry with Aldefluor™ assay in cells, isolated from human osteoarthritic (OA) cartilage. Its coexpression with surface markers was identified. Cells were sorted according to ALDH activity, and gene expression in sorted populations (ALDH(+) and ALDH(-)) was analyzed by RTq-PCR with Taqman(®) assay. RESULTS About 40% of freshly isolated chondrocytes demonstrated ALDH activity that remarkably declined during monolayer culture. Markers CD54 and CD55 were significantly stronger expressed, while CD47, CD140b, CD146 and CD166 were depleted in ALDH-expressing (ALDH(pos)) cells. Gene expression analysis revealed significantly higher expression of chondrocyte-specific genes COL2A1, SOX9 and SERPINA1 and lower expression of osteogenic markers RUNX2 and osteocalcin (BGLAP) in sorted ALDH(+) fraction. COL1A1, ACAN, ALPL and stem cell markers NANOG, OCT4, SOX2 and ABCG2 did not differ remarkably between the populations. Genes of isoenzymes ALDH1A2, ALDH1A3 and ALDH2 were strongly expressed, while ALDH1A1 was weakly expressed in chondrocytes. Only ALDH1A2 and ALDH1A3 were significantly enriched in ALDH(+) fraction. CONCLUSIONS We identified ALDH activity with significantly stronger expression of CD54 and CD55 in human articular chondrocytes. Gene expression of isotypes ALDH1A2, ALDH1A3 and ALDH2 was identified. Coexpression of ALDH activity with chondrogenic markers suggests its association with collagen II producing chondrocyte phenotype. Isotypes ALDH1A2 and ALDH1A3 can be associated with the ALDH activity in these cells.
Collapse
|
28
|
Xu Y, Lee J, Lü ZR, Mu H, Zhang Q, Park YD. Integration of Inhibition Kinetics and Molecular Dynamics Simulations: A Urea-Mediated Folding Study on Acetaldehyde Dehydrogenase 1. Appl Biochem Biotechnol 2016; 179:1101-14. [PMID: 27000059 DOI: 10.1007/s12010-016-2052-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 03/14/2016] [Indexed: 11/21/2022]
Abstract
Understanding the mechanism of acetaldehyde dehydrogenase 1 (ALDH1) folding is important because this enzyme is directly involved in several types of cancers and other diseases. We investigated the urea-mediated unfolding of ALDH1 by integrating kinetic inhibition studies with computational molecular dynamics (MD) simulations. Conformational changes in the enzyme structure were also analyzed using intrinsic and 1-anilinonaphthalene-8-sulfonate (ANS)-binding fluorescence measurements. Kinetic studies revealed that the direct binding of urea to ALDH1 induces inactivation of ALDH1 in a manner of mixed-type inhibition. Tertiary structural changes associated with regional hydrophobic exposure of the active site were observed. The urea binding regions on ALDH1 were predicted by docking simulations and were partly shared with active site residues of ALDH1 and with interface residues of the oligomerization domain for tetramer formation. The docking results suggest that urea prevents formation of the ALDH1 normal shape for the tetramer state as well as entrance of the substrate into the active site. Our study provides insight into the structural changes that accompany urea-mediated unfolding of ALDH1 and the catalytic role associated with conformational changes.
Collapse
Affiliation(s)
- Yingying Xu
- Zhejiang Provincial Key Laboratory of Applied Enzymology, Yangtze Delta Region Institute of Tsinghua University, Jiaxing, 314006, People's Republic of China.,School of Preclinical Medicine, Beijing University of Chinese Medicine, 11 Beisanhuan Dong Road, Beijing, 100029, People's Republic of China
| | - Jinhyuk Lee
- Korean Bioinformation Center (KOBIC), Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-806, South Korea.,Department of Nanobiotechnology and Bioinformatics, University of Sciences and Technology, Daejeon, 305-350, South Korea
| | - Zhi-Rong Lü
- Zhejiang Provincial Key Laboratory of Applied Enzymology, Yangtze Delta Region Institute of Tsinghua University, Jiaxing, 314006, People's Republic of China
| | - Hang Mu
- Zhejiang Provincial Key Laboratory of Applied Enzymology, Yangtze Delta Region Institute of Tsinghua University, Jiaxing, 314006, People's Republic of China
| | - Qian Zhang
- School of Preclinical Medicine, Beijing University of Chinese Medicine, 11 Beisanhuan Dong Road, Beijing, 100029, People's Republic of China.
| | - Yong-Doo Park
- Zhejiang Provincial Key Laboratory of Applied Enzymology, Yangtze Delta Region Institute of Tsinghua University, Jiaxing, 314006, People's Republic of China. .,College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo, 315100, People's Republic of China.
| |
Collapse
|
29
|
Park SJ, Yi B, Lee HS, Oh WY, Na HK, Lee M, Yang M. To quit or not: Vulnerability of women to smoking tobacco. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, ENVIRONMENTAL CARCINOGENESIS & ECOTOXICOLOGY REVIEWS 2016; 34:33-56. [PMID: 26669465 DOI: 10.1080/10590501.2015.1131539] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Tobacco smoking is currently on the rise among women, and can pose a greater health risk. In order to understand the nature of the increase in smoking prevalence among women, we focused on the vulnerability of women to smoking behaviors--smoking cessation or tobacco addiction--and performed a systematic review of the socioeconomic and intrinsic factors as well as tobacco ingredients that affect women's susceptibility to smoking tobacco. We observed that nicotine and other tobacco components including cocoa-relatives, licorice products, and menthol aggravate tobacco addiction in women rather than in men. Various genetic and epigenetic alterations in dopamine pathway and the pharmaco-kinetics and -dynamic factors of nicotine also showed potential evidences for high susceptibility to tobacco addiction in women. Therefore, we suggest systemic approaches to prevent tobacco smoking-related health risks, considering gene-environment-gender interaction.
Collapse
Affiliation(s)
- Se-Jung Park
- a Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University , Seoul , Republic of Korea
| | - Bitna Yi
- b Department of Neurosurgery , Stanford University School of Medicine , Stanford , California , USA
| | - Ho-Sun Lee
- a Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University , Seoul , Republic of Korea
| | - Woo-Yeon Oh
- a Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University , Seoul , Republic of Korea
| | - Hyun-Kyung Na
- a Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University , Seoul , Republic of Korea
| | - Minjeong Lee
- a Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University , Seoul , Republic of Korea
| | - Mihi Yang
- a Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University , Seoul , Republic of Korea
| |
Collapse
|
30
|
Takitani K, Inoue K, Koh M, Miyazaki H, Inoue A, Kishi K, Tamai H. Altered retinol status and expression of retinol-related proteins in streptozotocin-induced type 1 diabetic model rats. J Clin Biochem Nutr 2015. [PMID: 26060349 DOI: 10.3164/jcbn.14.113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Diabetes is a metabolic disorder characterized by chronic hyperglycemia. Advanced diabetes is associated with severe complications and impaired nutritional status. Here, we assessed the expression of retinol-associated proteins, including β-carotene 15,15'-monooxygenase (BCMO), lecithin:retinol acyltransferase (LRAT), aldehyde dehydrogenase (ALDH), and cytochrome P450 26A1 (CYP26A1), and measured retinol levels in the plasma and liver of streptozotocin (STZ)-induced type 1 diabetic model rats. Compared to the levels in the control rats, retinol levels in the plasma and liver of STZ rats were decreased and increased, respectively. Hepatic expression of the LRAT gene in STZ rats was lower than that in the controls. In the liver of STZ rats, the expression of ALDH1A1, a retinal metabolizing enzyme was higher, whereas ALDH1A2 expression was lower than in the controls. Hepatic CYP26A1 expression in STZ rats was significantly higher than in the control rats. BCMO expression levels in the liver and intestine of STZ rats were much lower than those of the controls. Altered BCMO expression might affect retinol status. It is considered that the metabolic availability of retinol was lessened despite the accelerated catabolism of retinol; therefore, retinol mobilization may be unbalanced in the liver of rats in the type 1 diabetic state.
Collapse
Affiliation(s)
- Kimitaka Takitani
- Department of Pediatrics, Osaka Medical College, 2-7 Daigakumachi, Takatsuki-shi, Osaka 569-8686, Japan
| | - Keisuke Inoue
- Department of Pediatrics, Hirakata City Hospital, 2-14-1 Kinyahonmachi, Hirakata-shi, Osaka 573-1013, Japan
| | - Maki Koh
- Department of Pediatrics, Osaka Medical College, 2-7 Daigakumachi, Takatsuki-shi, Osaka 569-8686, Japan
| | - Hiroshi Miyazaki
- Department of Pediatrics, Osaka Rosai Hospital, 1179-3 Nagasonecho, Kita-ku, Sakai-shi, Osaka 591-8025, Japan
| | - Akiko Inoue
- Department of Pediatrics, Osaka Medical College, 2-7 Daigakumachi, Takatsuki-shi, Osaka 569-8686, Japan
| | - Kanta Kishi
- Department of Pediatrics, Osaka Medical College, 2-7 Daigakumachi, Takatsuki-shi, Osaka 569-8686, Japan
| | - Hiroshi Tamai
- Department of Pediatrics, Osaka Medical College, 2-7 Daigakumachi, Takatsuki-shi, Osaka 569-8686, Japan
| |
Collapse
|
31
|
Kim YK, Zuccaro MV, Costabile BK, Rodas R, Quadro L. Tissue- and sex-specific effects of β-carotene 15,15' oxygenase (BCO1) on retinoid and lipid metabolism in adult and developing mice. Arch Biochem Biophys 2015; 572:11-18. [PMID: 25602705 PMCID: PMC4402122 DOI: 10.1016/j.abb.2015.01.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 12/30/2014] [Accepted: 01/02/2015] [Indexed: 12/20/2022]
Abstract
In mammals, β-carotene-15,15'-oxygenase (BCO1) is the main enzyme that cleaves β-carotene, the most abundant vitamin A precursor, to generate retinoids (vitamin A derivatives), both in adult and developing tissues. We previously reported that, in addition to this function, BCO1 can also influence the synthesis of retinyl esters, the storage form of retinoids, in the mouse embryo at mid-gestation. Indeed, lack of embryonic BCO1 impaired both lecithin-dependent and acyl CoA-dependent retinol esterification, mediated by lecithin:retinol acyltransferase (LRAT) and acyl CoA:retinol acyltransferase (ARAT), respectively. Furthermore, embryonic BCO1 also influenced the ester pools of cholesterol and diacylglycerol. In this report, we gained novel insights into this alternative function of BCO1 by investigating whether BCO1 influenced embryonic retinoid and lipid metabolism in a tissue-dependent manner. To this end, livers and brains from wild-type and BCO1-/- embryos at mid-gestation were analyzed for retinoid and lipid content, as well as gene expression levels. We also asked whether or not the role of BCO1 as a regulator of lecithin- and acyl CoA-dependent retinol esterification was exclusively restricted to the developing tissues. Thus, a survey of retinol and retinyl ester levels in adult tissues of wild-type, BCO1-/-, LRAT-/- and LRAT-/-BCO1-/- mice was performed. We showed that the absence of BCO1 affects embryonic retinoid and lipid homeostasis in a tissue-specific manner and that retinyl ester formation is also influenced by BCO1 in a few adult tissues (pancreas, lung, heart and adipose) in a sex-dependent manner.
Collapse
Affiliation(s)
- Youn-Kyung Kim
- Department of Food Science and Rutgers Center for Lipid Research, Rutgers University, New Brunswick, NJ 08091, USA
| | - Michael V Zuccaro
- Department of Food Science and Rutgers Center for Lipid Research, Rutgers University, New Brunswick, NJ 08091, USA
| | - Brianna K Costabile
- Department of Food Science and Rutgers Center for Lipid Research, Rutgers University, New Brunswick, NJ 08091, USA
| | - Rebeka Rodas
- Department of Food Science and Rutgers Center for Lipid Research, Rutgers University, New Brunswick, NJ 08091, USA
| | - Loredana Quadro
- Department of Food Science and Rutgers Center for Lipid Research, Rutgers University, New Brunswick, NJ 08091, USA.
| |
Collapse
|
32
|
Takitani K, Inoue K, Koh M, Miyazaki H, Inoue A, Kishi K, Tamai H. Altered retinol status and expression of retinol-related proteins in streptozotocin-induced type 1 diabetic model rats. J Clin Biochem Nutr 2015; 56:195-200. [PMID: 26060349 PMCID: PMC4454085 DOI: 10.3164/jcbn.14-113] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 10/22/2014] [Indexed: 01/08/2023] Open
Abstract
Diabetes is a metabolic disorder characterized by chronic hyperglycemia. Advanced diabetes is associated with severe complications and impaired nutritional status. Here, we assessed the expression of retinol-associated proteins, including β-carotene 15,15'-monooxygenase (BCMO), lecithin:retinol acyltransferase (LRAT), aldehyde dehydrogenase (ALDH), and cytochrome P450 26A1 (CYP26A1), and measured retinol levels in the plasma and liver of streptozotocin (STZ)-induced type 1 diabetic model rats. Compared to the levels in the control rats, retinol levels in the plasma and liver of STZ rats were decreased and increased, respectively. Hepatic expression of the LRAT gene in STZ rats was lower than that in the controls. In the liver of STZ rats, the expression of ALDH1A1, a retinal metabolizing enzyme was higher, whereas ALDH1A2 expression was lower than in the controls. Hepatic CYP26A1 expression in STZ rats was significantly higher than in the control rats. BCMO expression levels in the liver and intestine of STZ rats were much lower than those of the controls. Altered BCMO expression might affect retinol status. It is considered that the metabolic availability of retinol was lessened despite the accelerated catabolism of retinol; therefore, retinol mobilization may be unbalanced in the liver of rats in the type 1 diabetic state.
Collapse
Affiliation(s)
- Kimitaka Takitani
- Department of Pediatrics, Osaka Medical College, 2-7 Daigakumachi, Takatsuki-shi, Osaka 569-8686, Japan
| | - Keisuke Inoue
- Department of Pediatrics, Hirakata City Hospital, 2-14-1 Kinyahonmachi, Hirakata-shi, Osaka 573-1013, Japan
| | - Maki Koh
- Department of Pediatrics, Osaka Medical College, 2-7 Daigakumachi, Takatsuki-shi, Osaka 569-8686, Japan
| | - Hiroshi Miyazaki
- Department of Pediatrics, Osaka Rosai Hospital, 1179-3 Nagasonecho, Kita-ku, Sakai-shi, Osaka 591-8025, Japan
| | - Akiko Inoue
- Department of Pediatrics, Osaka Medical College, 2-7 Daigakumachi, Takatsuki-shi, Osaka 569-8686, Japan
| | - Kanta Kishi
- Department of Pediatrics, Osaka Medical College, 2-7 Daigakumachi, Takatsuki-shi, Osaka 569-8686, Japan
| | - Hiroshi Tamai
- Department of Pediatrics, Osaka Medical College, 2-7 Daigakumachi, Takatsuki-shi, Osaka 569-8686, Japan
| |
Collapse
|
33
|
Opdenaker LM, Arnold KM, Pohlig RT, Padmanabhan JS, Flynn DC, Sims-Mourtada J. Immunohistochemical analysis of aldehyde dehydrogenase isoforms and their association with estrogen-receptor status and disease progression in breast cancer. BREAST CANCER-TARGETS AND THERAPY 2014; 6:205-9. [PMID: 25540596 PMCID: PMC4270298 DOI: 10.2147/bctt.s73674] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
In many types of tumors, especially breast tumors, aldehyde dehydrogenase (ALDH) activity has been used to identify cancer stem-like cells within the tumor. The presence and quantity of these cells are believed to predict the response of tumors to chemotherapy. Therefore, identification and eradication of these cells would be necessary to cure the patient. However, there are 19 different ALDH isoforms that could contribute to the enzyme activity. ALDH1A1 and ALDH1A3 are among the isoforms mostly responsible for the increased ALDH activity observed in these stem-like cells, although the main isoforms vary in different tissues and tumor types. In the study reported here, we attempted to determine if ALDH1A1 or ALDH1A3, specifically, correlate with tumor stage, grade, and hormone-receptor status in breast-cancer patients. While there was no significant correlation between ALDH1A1 and any of the parameters tested, we were able to identify a positive correlation between ALDH1A3 and tumor stage in triple-negative cancers. In addition, ALDH1A3 was negatively correlated with estrogen-receptor status. Our data suggest that ALDH1A3 could be utilized as a marker to identify stem-like cells within triple-negative tumors.
Collapse
Affiliation(s)
- Lynn M Opdenaker
- Center for Translational Cancer Research, Helen F Graham Cancer Center, Christiana Care Health Services, Inc., Newark, Delaware, USA ; Department of Biological Sciences, University of Delaware, Newark, Delaware, USA
| | - Kimberly M Arnold
- Center for Translational Cancer Research, Helen F Graham Cancer Center, Christiana Care Health Services, Inc., Newark, Delaware, USA ; Department of Medical Laboratory Sciences, University of Delaware, Newark, Delaware, USA
| | - Ryan T Pohlig
- Department of Medical Laboratory Sciences, University of Delaware, Newark, Delaware, USA ; Biostatistics Core Facility, University of Delaware, Newark, Delaware, USA
| | - Jayasree S Padmanabhan
- Center for Translational Cancer Research, Helen F Graham Cancer Center, Christiana Care Health Services, Inc., Newark, Delaware, USA
| | - Daniel C Flynn
- Center for Translational Cancer Research, Helen F Graham Cancer Center, Christiana Care Health Services, Inc., Newark, Delaware, USA ; Department of Medical Laboratory Sciences, University of Delaware, Newark, Delaware, USA
| | - Jennifer Sims-Mourtada
- Center for Translational Cancer Research, Helen F Graham Cancer Center, Christiana Care Health Services, Inc., Newark, Delaware, USA ; Department of Biological Sciences, University of Delaware, Newark, Delaware, USA ; Department of Medical Laboratory Sciences, University of Delaware, Newark, Delaware, USA
| |
Collapse
|
34
|
DiSilvestro D, Petrosino J, Aldoori A, Melgar-Bermudez E, Wells A, Ziouzenkova O. Enzymatic intracrine regulation of white adipose tissue. Horm Mol Biol Clin Investig 2014; 19:39-55. [PMID: 25390015 DOI: 10.1515/hmbci-2014-0019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Accepted: 05/28/2014] [Indexed: 11/15/2022]
Abstract
Abdominal fat formation has become a permanent risk factor for metabolic syndrome and various cancers in one-third of the world's population of obese and even lean patients. Formation of abdominal fat involves additional mechanisms beyond an imbalance in energy intake and expenditure, which explains systemic obesity. In this review, we briefly summarized autonomous regulatory circuits that locally produce hormones from inactive precursors or nutrients for intra-/auto-/paracrine signaling in white adipose depots. Enzymatic pathways activating steroid and thyroid hormones in adipose depots were compared with enzymatic production of retinoic acid from vitamin A. We discussed the role of intracrine circuits in fat-depot functions and strategies to reduce abdominal adiposity through thermogenic adipocytes with interrupted generation of retinoic acid.
Collapse
|
35
|
Barber T, Esteban-Pretel G, Marín MP, Timoneda J. Vitamin a deficiency and alterations in the extracellular matrix. Nutrients 2014; 6:4984-5017. [PMID: 25389900 PMCID: PMC4245576 DOI: 10.3390/nu6114984] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 09/26/2014] [Accepted: 10/20/2014] [Indexed: 12/13/2022] Open
Abstract
Vitamin A or retinol which is the natural precursor of several biologically active metabolites can be considered the most multifunctional vitamin in mammals. Its deficiency is currently, along with protein malnutrition, the most serious and common nutritional disorder worldwide. It is necessary for normal embryonic development and postnatal tissue homeostasis, and exerts important effects on cell proliferation, differentiation and apoptosis. These actions are produced mainly by regulating the expression of a variety of proteins through transcriptional and non-transcriptional mechanisms. Extracellular matrix proteins are among those whose synthesis is known to be modulated by vitamin A. Retinoic acid, the main biologically active form of vitamin A, influences the expression of collagens, laminins, entactin, fibronectin, elastin and proteoglycans, which are the major components of the extracellular matrix. Consequently, the structure and macromolecular composition of this extracellular compartment is profoundly altered as a result of vitamin A deficiency. As cell behavior, differentiation and apoptosis, and tissue mechanics are influenced by the extracellular matrix, its modifications potentially compromise organ function and may lead to disease. This review focuses on the effects of lack of vitamin A in the extracellular matrix of several organs and discusses possible molecular mechanisms and pathologic implications.
Collapse
Affiliation(s)
- Teresa Barber
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universitat de Valencia, Avda V. Andrés Estellés s/n, 46100-Burjassot, Spain.
| | - Guillermo Esteban-Pretel
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universitat de Valencia, Avda V. Andrés Estellés s/n, 46100-Burjassot, Spain.
| | - María Pilar Marín
- Unidad de Microscopía IIS La Fe Valencia, Avda Campanar, 21, 46009-Valencia, Spain.
| | - Joaquín Timoneda
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universitat de Valencia, Avda V. Andrés Estellés s/n, 46100-Burjassot, Spain.
| |
Collapse
|
36
|
Weber TJ, Magnaldo T, Xiong Y. ALDH1A1 Deficiency in Gorlin Syndrome Suggests a Central Role for Retinoic Acid and ATM Deficits in Radiation Carcinogenesis. Proteomes 2014; 2:451-467. [PMID: 28250390 PMCID: PMC5302750 DOI: 10.3390/proteomes2030451] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 08/11/2014] [Accepted: 09/01/2014] [Indexed: 12/14/2022] Open
Abstract
We hypothesize that aldehyde dehydrogenase 1A1 (ALDH1A1) deficiency will result in impaired ataxia-telangiectasia mutated (ATM) activation in a retinoic acid-sensitive fashion. Data supporting this hypothesis include (1) reduced ATM activation in irradiated primary dermal fibroblasts from ALDH1A1-deficient Gorlin syndrome patients (GDFs), relative to ALDH1A1-positive normal human dermal fibroblasts (NHDFs) and (2) increased ATM activation by X-radiation in GDFs pretreated with retinoic acid, however, the impact of donor variability on ATM activation in fibroblasts was not assessed and is a prudent consideration in future studies. Clonogenic survival of irradiated cells showed differential responses to retinoic acid as a function of treatment time. Long-term (5 Day) retinoic acid treatment functioned as a radiosensitizer and was associated with downregulation of ATM protein levels. Short-term (7 h) retinoic acid treatment showed a trend toward increased survival of irradiated cells and did not downregulate ATM protein levels. Using a newly developed IncubATR technology, which defines changes in bulk chemical bond patterns in live cells, we can discriminate between the NHDF and GDF phenotypes, but treatment of GDFs with retinoic acid does not induce reversion of bulk chemical bond patterns associated with GDFs toward the NHDF phenotype. Collectively, our preliminary investigation of the Gorlin phenotype has identified deficient ALDH1A1 expression associated with deficient ATM activation as a possible susceptibility factor that is consistent with the high incidence of spontaneous and radiation-induced carcinogenesis in these patients. The IncubATR technology exhibits sufficient sensitivity to detect phenotypic differences in live cells that may be relevant to radiation health effects.
Collapse
Affiliation(s)
- Thomas J Weber
- Systems Toxicology and Exposure Science, Pacific Northwest National Laboratory, Richland, WA 99352, USA.
| | - Thierry Magnaldo
- Faculté de Médicine, 2ème étage, CNRS UMR 6267-INSERM U998-UNSA, Nice 06107 Cedex 2, France.
| | - Yijia Xiong
- College of Osteopathic Medicine of the Pacific-Northwest, Western University of Health Sciences, Lebanon, OR 97355, USA.
| |
Collapse
|